the coincidence between increasing age, immunosuppression, … · 2020. 10. 1. · (wick et al.,...

13
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 ORIGINAL RESEARCH published: xx February 2019 doi: 10.3389/fphar.2019.00200 Edited by: Andrew Zloza, Rush University Medical Center, United States Reviewed by: Praveen Bommareddy, Rutgers, The State University of New Jersey, United States Tomas Garzon-Muvdi, Thomas Jefferson University Hospitals, United States *Correspondence: Derek A. Wainwright [email protected] Specialty section: This article was submitted to Cancer Molecular Targets and Therapeutics, a section of the journal Frontiers in Pharmacology Received: 12 January 2019 Accepted: 18 February 2019 Published: xx February 2019 Citation: Ladomersky E, Scholtens DM, Kocherginsky M, Hibler EA, Bartom ET, Otto-Meyer S, Zhai L, Lauing KL, Choi J, Sosman JA, Wu JD, Zhang B, Lukas RV and Wainwright DA (2019) The Coincidence Between Increasing Age, Immunosuppression, and the Incidence of Patients With Glioblastoma. Front. Pharmacol. 10:200. doi: 10.3389/fphar.2019.00200 The Coincidence Between Increasing Age, Immunosuppression, and the Incidence of Patients With Glioblastoma Erik Ladomersky 1 , Denise M. Scholtens 1,2 , Masha Kocherginsky 2,3 , Elizabeth A. Hibler 2 , Elizabeth T. Bartom 4 , Sebastian Otto-Meyer 1 , Lijie Zhai 1 , Kristen L. Lauing 1 , Jaehyuk Choi 4,5 , Jeffrey A. Sosman 6 , Jennifer D. Wu 7,8 , Bin Zhang 6,8 , Rimas V. Lukas 9,10 and Derek A. Wainwright 1,6,8,9 * 1 Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 2 Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 3 Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 4 Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 5 Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 6 Department of Medicine-Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 7 Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 8 Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 9 Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States, 10 Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States Background: Glioblastoma (GBM) is the most aggressive primary brain tumor in adults and is associated with a median overall survival (mOS) of 16–21 months. Our previous work found a negative association between advanced aging and the survival benefit after treatment with immunotherapy in an experimental brain tumor model. Given the recent phase III clinical success of immunotherapy in patients with many types of cancer, but not for patients with GBM, we hypothesize that aging enhances immunosuppression in the brain and contributes to the lack of efficacy for immunotherapy to improve mOS in patients with malignant glioma. Herein, we compare epidemiological data for the incidence and mortality of patients with central nervous system (CNS) cancers, in addition to immune-related gene expression in the normal human brain, as well as peripheral blood immunological changes across the adult lifespan. Methods: Data were extracted from the National Cancer Institute’s surveillance, epidemiology, and end results (SEER)-, the Broad Institute’s Genotype Tissue Expression project (GTEx)-, and the University of California San Francisco’s 10k Immunomes-databases and analyzed for associations with aging. Results: The proportion of elderly individuals, defined as 65 years of age, has predominantly increased for more than 100 years in the United States. Over time, the rise in elderly United States citizens has correlated with an increased incidence and mortality rate associated with primary brain and other CNS cancer. With advanced aging, human mRNA expression for factors associated with immunoregulation including immunosuppressive indoleamine 2,3 dioxygenase 1 (IDO) and programmed Frontiers in Pharmacology | www.frontiersin.org 1 February 2019 | Volume 10 | Article 200

Upload: others

Post on 10-Nov-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 1

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

ORIGINAL RESEARCHpublished: xx February 2019

doi: 10.3389/fphar.2019.00200

Edited by:Andrew Zloza,

Rush University Medical Center,United States

Reviewed by:Praveen Bommareddy,

Rutgers, The State Universityof New Jersey, United States

Tomas Garzon-Muvdi,Thomas Jefferson University

Hospitals, United States

*Correspondence:Derek A. Wainwright

[email protected]

Specialty section:This article was submitted to

Cancer Molecular Targetsand Therapeutics,

a section of the journalFrontiers in Pharmacology

Received: 12 January 2019Accepted: 18 February 2019Published: xx February 2019

Citation:Ladomersky E, Scholtens DM,

Kocherginsky M, Hibler EA,Bartom ET, Otto-Meyer S, Zhai L,

Lauing KL, Choi J, Sosman JA,Wu JD, Zhang B, Lukas RV and

Wainwright DA (2019) TheCoincidence Between Increasing Age,

Immunosuppression,and the Incidence of Patients With

Glioblastoma.Front. Pharmacol. 10:200.

doi: 10.3389/fphar.2019.00200

The Coincidence Between IncreasingAge, Immunosuppression, and theIncidence of Patients WithGlioblastomaErik Ladomersky1, Denise M. Scholtens1,2, Masha Kocherginsky2,3, Elizabeth A. Hibler2,Elizabeth T. Bartom4, Sebastian Otto-Meyer1, Lijie Zhai1, Kristen L. Lauing1,Jaehyuk Choi4,5, Jeffrey A. Sosman6, Jennifer D. Wu7,8, Bin Zhang6,8, Rimas V. Lukas9,10

and Derek A. Wainwright1,6,8,9*

1 Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States,2 Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States,3 Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL,United States, 4 Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine,Chicago, IL, United States, 5 Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL,United States, 6 Department of Medicine-Hematology and Oncology, Northwestern University Feinberg School of Medicine,Chicago, IL, United States, 7 Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL,United States, 8 Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago,IL, United States, 9 Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine,Chicago, IL, United States, 10 Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL,United States

Background: Glioblastoma (GBM) is the most aggressive primary brain tumor in adultsand is associated with a median overall survival (mOS) of 16–21 months. Our previouswork found a negative association between advanced aging and the survival benefit aftertreatment with immunotherapy in an experimental brain tumor model. Given the recentphase III clinical success of immunotherapy in patients with many types of cancer, butnot for patients with GBM, we hypothesize that aging enhances immunosuppressionin the brain and contributes to the lack of efficacy for immunotherapy to improvemOS in patients with malignant glioma. Herein, we compare epidemiological data forthe incidence and mortality of patients with central nervous system (CNS) cancers,in addition to immune-related gene expression in the normal human brain, as well asperipheral blood immunological changes across the adult lifespan.

Methods: Data were extracted from the National Cancer Institute’s surveillance,epidemiology, and end results (SEER)-, the Broad Institute’s Genotype TissueExpression project (GTEx)-, and the University of California San Francisco’s 10kImmunomes-databases and analyzed for associations with aging.

Results: The proportion of elderly individuals, defined as ≥65 years of age, haspredominantly increased for more than 100 years in the United States. Over time, therise in elderly United States citizens has correlated with an increased incidence andmortality rate associated with primary brain and other CNS cancer. With advancedaging, human mRNA expression for factors associated with immunoregulationincluding immunosuppressive indoleamine 2,3 dioxygenase 1 (IDO) and programmed

Frontiers in Pharmacology | www.frontiersin.org 1 February 2019 | Volume 10 | Article 200

Page 2: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 2

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

death-ligand 1 (PD-L1), as well as the dendritic cell surface marker, CD11c,increase in the brain of normal human subjects, coincident with increased circulatingimmunosuppressive Tregs and decreased cytolytic CD8+ T cells in the peripheral blood.Strikingly, these changes are maximally pronounced in the 60–69 year old group;consistent with the median age of a diagnosis for GBM.

Conclusion: These data demonstrate a significant association between normal humanaging and increased immunosuppression in the circulation and CNS; particularly late inlife. Our data raise several hypotheses including that, aging: (i) progressively suppressesnormal immunosurveillance and thereby contributes to GBM cell initiation and/oroutgrowth; (ii) decreases immunotherapeutic efficacy against malignant glioma.

Keywords: aging, biomarker, IDO, immunosuppression, PD-L1, immunotherapy, Treg, IDH

INTRODUCTION

Glioblastoma (GBM) is the most common primary malignantbrain tumor in adults. Despite the aggressive standard of careregimen that includes maximal surgical resection followed byradiation therapy and chemotherapy with temozolomide, andmore recently tumor treating fields, the median overall survival(mOS) remains at 16–21 months post-diagnosis, with just43% of patients surviving for 2 years post-diagnosis (Stuppet al., 2005, 2009, 2017; Johnson and O’Neill, 2012). A majorfactor contributing to the poor GBM patient prognosis is thepotent immunosuppression, found systemically and locally inthe brain tumor microenvironment (Chongsathidkiet et al.,2018). High intratumoral expression of immunosuppressivemediators including programmed cell death protein-1 (PD-1)and indoleamine 2,3 dioxygenase 1 (IDO), is prognostic fordecreased GBM patient survival (Wainwright et al., 2012; Nduomet al., 2016; Zhai et al., 2017). Immune checkpoint inhibitortreatment has demonstrated a survival benefit in patients withnon-small cell lung carcinoma (Antonia et al., 2016), renal cellcancer (Motzer et al., 2015), end-stage melanoma (Larkin et al.,2015), and other aggressive malignancies arising outside of thecentral nervous system (CNS). In contrast, this benefit has yet totranslate into patients with GBM in phase III clinical trials to-date(Bristol-Myers Squibb, 2017; Filley et al., 2017).

Age is one of the primary risk factors for cancer, withindividuals ≥65 years of age accounting for 60% of newlydiagnosed malignancies and 70% of all cancer-related deaths(Ries et al., 2006). A similar report highlighted an age adjustedcancer mortality rate for persons ≥65 at ∼16 times higherthan the mortality rate for those <65 (Berger et al., 2006).Care for these individuals is challenging due to the number ofdiseases elderly subjects are at high risk for, which also raises thelikelihood of presenting multiple comorbidities during advancedaging (Yancik et al., 1998, 2001). Similarly, the incidence andmortality rate of GBM increases during advanced aging witha median diagnosis at 64 years old (Young et al., 2017).Aging is a complex process that affects nearly all aspectsof the immune system (Nikolich-Zugich, 2018). In general,advanced aging decreases immune system effectiveness, as isevidenced in elderly individuals who receive the influenza vaccine

(McElhaney and Dutz, 2008). Aging also negatively impactsapoptotic cell clearance (Aprahamian et al., 2008), the numbersof naïve T cells (Cambier, 2005), and the wound healing response(Wick et al., 2010). T cell senescence increases with progressiveaging through the induction of p16 (Liu et al., 2009). Aging alsoaffects T cell receptor (TCR) signaling in CD4+ T cells, duein part to decreased miR-181a; a microRNA highly expressedin normal T cells (Adachi and Davis, 2011; Li et al., 2012;Chen et al., 2013). Both aged mice (Rossi et al., 2005) andhuman (Pang et al., 2011) hematopoietic stem cells (HSC)possess a myeloid-biased differentiation potential as comparedwith HSC from young subjects. Moreover, macrophages fromdonor subjects with advanced age possess decreased capacityfor antigen presentation as compared to young donors (Davilaet al., 1990; Gon et al., 1996). Neuro-immune interactions arealso affected by aging in the brain (Carson et al., 2006) thatinclude a significant upregulation of MHCII and CD11b onmicroglia (Rogers et al., 1988; Perry et al., 1993), as well as anaccumulation of brain-resident dendritic cells (Bulloch et al.,2008; D’Agostino et al., 2012; Kaunzner et al., 2012). However,it is not yet clear as to whether aging possesses a distinct impacton immunosuppressive gene expression across select tissues thatcontribute to a microenvironment permissive for oncogenesis,tumor progression, and/or resistance to immunoregulatory-based therapies.

Our laboratory previously demonstrated a negative impactof advanced age on the survival of animals engrafted withsyngeneic experimental brain tumors (Ladomersky et al., 2016).C57BL/6 mice intracranially injected with murine GL261 gliomacells at 72–74 weeks old, which is similar in humans to atime frame associated with the median age of a GBM patientdiagnosis (Fox et al., 2007; Dutta and Sengupta, 2016), havea decreased mOS as compared to animal subjects with an ageof 6–8 weeks old (27.5 and 21.5 days, respectively, P = 0.029,n = 10–12/group); the latter of which is similar in age toa human teenager. The negative impact of advanced agingwas coincident with increased immunosuppressive IDO1 geneexpression in the normal, non-malignant mouse brain. Morerecently, we discovered that a substantial proportion of C57BL/6mice intracranially injected with GL261 at 6–8 weeks of ageexperience long-term survival when simultaneously treated with

Frontiers in Pharmacology | www.frontiersin.org 2 February 2019 | Volume 10 | Article 200

Page 3: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 3

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

radiation (RT), anti-PD-1 mAb, and IDO1 enzyme inhibitor(Ladomersky et al., 2018). The brain tumor survival benefitprovided by this treatment, however, was negatively affected byanimal subjects with advanced age as compared with youngsubjects (Ladomersky et al., 2018). Importantly, there was nosignificant difference in tumor infiltrating leukocyte populationsbetween the young and aged subjects within treatment groups. Toour knowledge, this is the first preclinical primary brain cancerstudy to demonstrate a negative impact of aging on survival aftertreatment with immunotherapy.

Further supporting the hypothesis that, advanced agingmediates suppression of immune system efficacy against atumor challenge event, previous work showed that splenocytesisolated from young, but not old immunized subjects, wereable to eradicate subcutaneous tumors in mice (Schreiber et al.,2012). Specifically, immunodeficient recombination activatinggene knockout mice (Rag−/−) were subcutaneously engrafted8101 cells arising from mice treated with UV-irradiation,and possessing a somatic mutation in the T cell-recognizedantigen RNA helicase, p68. Splenocytes isolated from 5 monthold mice and immunized with live 8101 cells, but notthose from immunized 29 month old mice, eradicated 8101cell-based tumors post-adoptive transfer into Rag−/− mice.Interestingly, melanoma patients≥62 years of age show increasedresponsiveness to anti-PD-1 mAb treatment as compared withyounger human subjects (Kugel et al., 2018). Recapitulating thisclinical observation, 10 month old animal subjects, which roughlycorrelate to the human age of 38–47 years and engrafted withmurine BSC9AJ2 melanoma cells, show decreased tumor growthas compared to 2 month old engrafted mice after treatment withanti-PD-1 mAb (Kugel et al., 2018). This highlights an interestingdichotomy suggesting that, the productivity of an anti-tumorimmune response during treatment with immunotherapy likelydepends on both the cancer type and age of the host. Thesecombined findings may suggest that GBM is an outlier whenconsidering its place in cancer immunology and immunotherapy.Accordingly, we previously found an inverse association betweenhigh CD3ε and CD8α gene expression with GBM patient survival(Zhai et al., 2017), which is a diametrically opposite finding ascompared with non-small cell lung cancer and melanoma (Zenget al., 2016; Zhai et al., 2018).

In our current study, we explored the associations betweenhuman: (i) aging; (ii) levels of gene expression associatedwith immunoregulation inside the brain; (iii) immunologicalchanges in the peripheral blood; and (iv) incidence andmortality of patients with primary brain and other CNS tumors.Epidemiological analyses of GBM patient characteristics werecompared across the Surveillance, Epidemiology, and End Results(SEER) database, age-dependent gene expression levels of normalhuman brain from the GTEx database, and age-associatedchanges in normal human peripheral blood leukocytes fromthe 10k Immunomes database. Our study confirms the strikingobservation that the brain cancer mortality rate is actively rising,with a particular enrichment among the elderly population inthe United States. The incidence of GBM is 3.4× higher amongindividuals ≥65 years old, as compared to those <65, whilethe mortality rate for individuals ≥65 years old with GBM is

7× higher as compared to GBM patients <65. Gene expressionlevels of immunosuppressive IDO1 increased in the normalhuman brain and was maximal among individuals aged 60–69 years old. As compared to younger subjects, there was amaximal incidence of circulating immunosuppressive regulatoryT cells (Tregs) and a significantly decreased cytolytic CD8+ Tcell population among the 60–69 year old age group. Our studyfound a cumulative peak index for immunosuppressive and/orimmunoregulatory mediators during the time frame associatedwith the median age of a GBM patient diagnosis. These dataraise the intriguing possibility that aging suppresses mechanismsof immunosurveillance and responsiveness to immunotherapy,which is associated with the increasing number of elderly patientswith brain cancer and the failure of immunotherapy to benefitGBM patients in phase III clinical trials to-date, respectively.

MATERIALS AND METHODS

DataLife Expectancy DataLife expectancy data were analyzed from the Center for DiseaseControl and Prevention. Data were accessed through the NationalCenter for Health Statistics portal1. The filename used was Lifeexpectancy at birth and at age 65, and at age 75, by sex, race, andHispanic origin: United States, selected years 1900–2016.

Surveillance of Epidemiology and End ResultsDatabase (SEER)All population, incidence, and mortality data from the SEERdatabase were accessed through SEER∗Stat (Version 8.3.52).Population-level data were accessed through a Frequency Session.Variables examined were: (1) Age recode with <1 year olds; and(2) year. Incidence and mortality data were accessed through aRate Session. Variables examined for incidence include: (1) agerecode with <1 year olds; (2) year of diagnosis; (3) histologyrecode – broad groupings; (4) histology recode – brain groupings;and (5) COD to site recode. The COD to site recode was usedto analyze the mortality rate of GBM. Variables examined formortality include: (1) age recode with <1 year old; (2) yearof death; and (3) cause of death recode. All rate data werecrude/non-age-adjusted. Data were accessed on 12-05-2018.

NCI 2018 EstimatesEstimated incidence and mortality data were retrieved from theNCI Cancer Stat Facts website3. The top 16 causes of cancer wereanalyzed for both incidence and mortality. Comparison acrossthe 16 cancers for the mortality to incidence ratio estimate werecalculated by dividing the estimated mortality with the estimatedincidence in the year 2018. Cost data were accessed through theNCI Cancer Prevalence and Cost of Care Projections website4.Cost per patient was calculated by taking the 2018 estimated

1https://www.cdc.gov/nchs/hus/contents2017.htm#0152https://seer.cancer.gov/seerstat/3https://seer.cancer.gov/statfacts/html/common.html4https://costprojections.cancer.gov/graph.php#

Frontiers in Pharmacology | www.frontiersin.org 3 February 2019 | Volume 10 | Article 200

Page 4: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 4

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

total charges for each type of cancer and dividing it by the 2018estimated incidence of each cancer type.

TCGASurvival data for GBM patients were analyzed from the cancergenome atlas. The data were accessed using the UCSC Xenaportal. Data were accessed on 02-01-2019.

10k ImmunomesNormal human peripheral blood cell data were analyzed from the10k Immunomes database. The data were accessed through theUCSF portal5. All data analyzed were CyTOF data plotted by age.Data were accessed on 06-22-2018.

GTExGene expression data for normal human tissues were analyzedfrom the GTEx database. Data were accessed through the dbGaPportal. All data were represented as log-transformed fragmentsper kilobase of transcript per million mapped reads (FPKM).These data were then plotted by subject age.

Statistical Analysis10k Immunomes and TCGA expression data are represented asthe mean ± SEM. GTEx data are represented by each individualvalue. The statistical significance of differences in cell counts(CyTOF) and gene expression (log-transformed FPKM) betweentwo groups was determined by Student t-test. Differences amongmultiple groups were assessed using ANOVA with post-hocTukeytest. The statistical significance of TCGA survival data wasdetermined by Log-rank test. Data were analyzed using Prismsoftware (GraphPad Software). A P-value less than 0.05 wasconsidered significant.

RESULTS

The Rate of Cancer Incidence IncreasesWith AgeOver the past 40 years in the United States, improved healthcaresystems and enhanced awareness of proper nutrition and exercisehave led to substantial increases in mean life expectancy, risingfrom 72.6 years of age in 1975, to 78.8 years of age in 2015(Figure 1A). Coincident with the increasing life expectancy in theUnited States, the proportion of elderly (≥65 years) individualsis rising. The elderly age group represented 10.6% of the totalpopulation in 1975, and increased to 14.9% in 2015 (Figure 1B).Both the incidence and mortality rates associated with cancerdiagnoses are higher among the elderly population as comparedto individuals <65 years of age (Figures 1C,D). In the year 1975,the incidence and mortality rate within the elderly populationfor all malignancies was 1,732/100,000 people and 942/100,000,respectively, whereas in the year 2015, the incidence rate for allmalignancies in the elderly population was 1,876/100,000 andthe mortality rate was 879/100,000. Compared to the population<65, these incidence and mortality rates were 9.6 and 12.1

5http://10kimmunomes.ucsf.edu/

FIGURE 1 | Progressive aging is associated with an increased rate of cancerincidence and mortality. (A) Life expectancy from the Center for DiseaseControl and Prevention (CDC) database for an average lifespan in theUnited States between the years 1975 and 2015. (B) Population data fromthe Surveillance, Epidemiology, and Ends Results (SEER) databaserepresenting the total number of elderly human subjects (ages ≥65) divided bythe total population in the United States for the years 1975, 1995, and 2015.(C) Incidence and (D) mortality rates from the SEER database for allmalignancies in the years 1975, 1995, and 2015. Rates are defined asnumber of cases and deaths, respectively, divided by the population in eachage category and multiplied by 100,000 people (per 100,000).

times higher, respectively, in 1975, and 7.1 and 13.6 timeshigher, respectively, in 2015. The slight decrease in mortalityrate between 1975 and 2015 is most likely due to improveddetection and treatment techniques in select cancers. However,due to the increasing elderly population, the absolute numbersof cancer related mortalities in this population has risen from214,173 in 1975, to 419,389 in 2015. Together, these data suggestan association between the aging population and an absoluteincrease of cancer-related deaths in the elderly population ofthe United States.

Brain Cancer Diagnoses Pose a GrowingChallenge to the United StatesHealthcare SystemPrimary brain tumors arising from a transformed cell within theCNS is a relatively rare form of cancer, with the 16th highestrate of incidence among all cancers, and an estimated 23,880 newpatient diagnoses in 2018 (Figure 2A). Dwarfing this numberis the estimated 266,120 new diagnoses for patients with breastcancer in the year 2018. The overall ratio of estimated breast

Frontiers in Pharmacology | www.frontiersin.org 4 February 2019 | Volume 10 | Article 200

Page 5: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 5

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

cancer incidence, as compared with brain cancer, is 11:1. Incontrast, the estimated mortality rate for patients diagnosed withCNS cancer is 16,830 in 2018 (Figure 2B), and is 40,920 forindividuals diagnosed with breast cancer. The overall ratio ofbreast cancer-associated mortalities, as compared to mortalitydue to CNS cancer, is only 2.4:1. Aggressiveness of cancerdiagnoses can also be calculated with the mortality-to-incidenceratio (MIR), which is determined by dividing overall mortalitywith the number of new diagnoses in a given year (Choi et al.,2017). CNS cancer as a group possesses the third highest MIR of0.70, which is only exceeded by pancreatic and liver cancer, withMIR scores of 0.80 and 0.72, respectively (Figure 2C). Leading allother groups, treatment for CNS cancers is the most costly on aper patient basis, with an average cost estimated to be $225,364 in2018 (Figure 2D).

Brain Cancer Incidence/Mortality IsRising and Enriched in the ElderlyAlthough there has only been a small change in the incidenceand mortality rates of all malignancies (Figures 1C,D) from 1975

FIGURE 2 | Brain andQ1 other CNS cancer is associated with a highmortality/incidence ratio and is expensive to treat. The estimated total (A)incidence and (B) mortality rate for the top 16 cancer types according to NCICancer Stat Facts. (C) The calculated mortality to incidence ratio (estimatedmortality/estimated incidence) for the top 16 cancer sites according to NCICancer Stat Facts. The dotted line represents an average across all 16 cancertypes. (D) Cost (United States dollars) per patient for each of the top 16cancer types. Dotted line represents the average for all 16 cancer types. NR,Not reported.

to 2015, analysis of incidence data for brain and other CNScancer shows a different trend (Figure 3A). The incidence ratehas increased overall from 5.4/100,000 in 1975, to 7.0/100,000 in2015. The overall increase, however, is primarily attributable tothe elderly population. In 1975 the incidence rate for individualswith brain and other CNS malignancies ≥65 years of age was14.2/100,000, which increased nearly 37% to 19.4/100,000 in2015. In that same time span, the incidence rate for those<65 years of age only slightly increased from 4.4/100,000 in1975 to 5.0/100,000 in 2015. When comparing other malignancytypes including breast, pancreatic, and lung, a similar trendof increased incidence was observed. Breast cancer incidencerates increased from 47.4/100,000 to 78.0/100,000 in the totalpopulation over the same time span, and from 192.4/100,000to 242.6/100,000 among the elderly population (Figure 3B).Pancreatic cancer incidence rates rose more slightly, from9.4/100,000 to 14.5/100,000 in the total population and from63.3/100,000 to 68.8/100,000 among the elderly over the past40 years (Figure 3C). The incidence of melanoma increased from6.8/100,000 to 28.4/100,000 in the total population and from16.7/100,000 to 97.7/100,000 within the elderly population from1975 to 2015 (Figure 3D). Overall, more people were diagnosedwith malignancies in 2015 as compared with 1975.

FIGURE 3 | Incidence rate for brain, breast, pancreatic, and melanomamalignancies over time. Incidence rates for (A) brain and other CNS, (B)breast, (C) pancreatic, and (D) melanoma malignancies analyzed from theSEER database for the years 1975, 1995, and 2015. Rates defined asnumber of cases divided by the population in each age category multiplied by100,000 people (per 100,000).

Frontiers in Pharmacology | www.frontiersin.org 5 February 2019 | Volume 10 | Article 200

Page 6: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 6

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

684

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

Similar to the increased rate of incidence, the overall mortalityrate among individuals with brain and other CNS cancersincreased in the total population from 3.8/100,000 in 1975to 5.1/100,000 in 2015 (Figure 4A) The mortality rate forindividuals with brain and other CNS malignancies≥65 years oldwas 11.6/100,000 in 1975, and increased by 56% to 18.1/100,000in 2015. In contrast, the mortality rate for individuals <65 yearsof age rose slightly from 2.8/100,000 in 1975 to 2.9/100,000 in2015. The overall mortality rate for individuals with breast cancerdecreased from 15.1/100,000 in 1975 to 13.1/100,000 in 2015, andfrom 66.8/100,000 to 52.9/100,000 among individuals ≥65 yearsof age during the same time periods, respectively (Figure 4B).The overall mortality rate for individuals with pancreatic cancerrose from 9.0/100,000 to 13/100,000 from 1975 to 2015, butonly increased slightly from 56.2/100,000 to 63.0/100,000 amongindividuals ≥65 years of age during the same time periods,respectively (Figure 4C). The overall mortality rate increased forpatients with melanoma from 1.8/100,000 to 2.8/100,000 from1975 to 2015 and increased from 6.2/100,000 in 1975 to 12.2 in2015 among individuals ≥65 years of age during the same timeperiods, respectively (Figure 4D).

FIGURE 4 | Mortality rate for brain, breast, pancreatic, and melanomamalignancies over time. Mortality rates for (A) brain and other CNS, (B)breast, (C) pancreatic, and (D) melanoma malignancies analyzed from theSEER database for the years 1975, 1995, and 2015. Rates defined asnumber of deaths divided by the population in each age category multipliedby 100,000 people (per 100,000).

When analyzing the MIR as a function of aging, there weredifferences in trends among the malignancies analyzed. While theoverall MIR for individuals with brain cancer increased slightlyfrom 0.70 in 1975 to 0.73 in 2015 (Figure 5A), there is a largerincrease from 0.82 in 1975 to 0.93 in 2015 among individuals≥65. In contrast, the overall MIR for individuals with breastcancer decreased over the same time span from 0.32 in 1975to 0.17 in 2015 (Figure 5B), with a similar decrease of 0.35in 1975 to 0.22 in 2015 among individuals ≥65. The overallMIR for individuals with pancreatic cancer was 0.96 in 1975and slightly decreased to 0.90 in 2015 (Figure 5C), whereas thisfigure is relatively unchanged within the elderly population at0.89 in 1975 to 0.90 in 2015. The overall MIR for individualswith melanoma decreased from 0.26 to 0.10 in 2015, and from0.37 to 0.12 among individuals ≥65, respectively (Figure 5D).Taken together, these data show substantially different trends ofaggressiveness, historically and currently, with brain cancer andother CNS cancers demonstrating a substantial increase in overallincidence, mortality, and MIR among the elderly portion of theUnited States population.

FIGURE 5 | Mortality to incidence ratio for brain, breast, pancreatic, andmelanoma malignancies over time. The calculated mortality to incidence ratioof (A) brain and other CNS, (B) breast, (C) pancreatic, and (D) melanomamalignancies for each year calculated from SEER incidence and mortalitydata. Instances where the ratio is over 1.0 are due to the lag time betweendiagnosis and mortality in GBM and pancreatic cancer.

Frontiers in Pharmacology | www.frontiersin.org 6 February 2019 | Volume 10 | Article 200

Page 7: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 7

685

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

702

703

704

705

706

707

708

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

726

727

728

729

730

731

732

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

750

751

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

777

778

779

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

The Incidence and Mortality ofIndividuals With GBM Is ProgressivelyEnriched With AgingGlioblastoma (GBM; grade IV) is the most common primarymalignant brain tumor in adults and accounts for 54% ofmalignant glioma diagnoses (Wen and Kesari, 2008). Amongtotal glioma incidence, the rate of a GBM diagnosis issubstantially higher as compared with grade II and grade IIIglioma (Figure 6A). The rate of a GBM patient diagnosis andmortality is enriched among the elderly population (Figure 6)and the MIR for individuals ≥65 years of age is 1.00.

Not only is GBM enriched among the elderly population,but the prognosis for these patients is substantially worse ascompared with individuals <65 years of age. We analyzed overallsurvival and expression data from the cancer genome atlas(TCGA) for individuals with GBM who possessed correspondingpatient data, expression data, and a reported IDH status (n = 144).There were only 8 patients within the dataset reporting thepresence of mutant IDH (mIDH), all of which were <65 yearsof age. There is a significantly shorter mOS of 11.3 monthsamong individuals≥65 years of age as compared with individuals<65 years of age (14.5 months; P = 0.019) (Figure 7A). Incontrast, mOS for the eight individuals with mIDH GBM is astriking 27.9 months. Intratumoral gene expression levels forthe immunosuppressive enzyme, IDO1, is similar for individuals<65 and ≥65 with wild-type IDH status, and significantlydecreased among mIDH GBM (Figures 7B,C) as was previously

FIGURE 6 | The incidence and mortality rates for glioblastoma (GBM) patientsare enriched among the elderly population. (A) SEER incidence rates (per100,000) for grade II glioma, grade III glioma, and GBM (grade IV glioma; allleft axis) compared to the percent of the total population represented for eachage group. Grade II and III glioma incidence rates calculated by SEER were>0.015 (per 100,000) for each age category. (B) The SEER mortality rate forGBM as compared to the percent of the total population represented for eachage group. Incidence and mortality data is represented as a summation forthe years 2011–2015.

FIGURE 7 | Prognosis is worse for individuals with GBM ≥65. Analysis ofGBM data for individuals with corresponding patient data, gene expression(HiSeq) data, and a reported mIDH1/2 status from the TCGA. (A) Survival dataof individuals with wild-type IDH1/2 <65 (n = 84), ≥65 (n = 52), and thosewith mIDH1/2 (n = 8). IDO1 gene expression (B) dot plot and (C) bar graph ofindividuals with wild-type IDH1/2 <65 (blue), ≥65 (red), and those withmIDH1/2 (green). (D) The percentage of GBM diagnoses within each agecategory for individuals with wtIDH1/2 (blue) and mIDH1/2 (green). (E) Survivaltime in days of individuals diagnosed in each age category. ∗P < 0.05.

reported (Zhai et al., 2017). While a majority of individuals withwild-type IDH GBM were diagnosed later in life, 50 percent ofindividuals with mIDH GBM were diagnosed before the age of35 (Figure 7D). Not surprisingly, overall survival time decreasedwith progressively increasing age among individuals with GBM(Figure 7E). Taken together, the data indicate that individualswith a wild-type IDH GBM, which constitute the majority of newdiagnoses, possess substantial comorbidity with advanced aging.

The Median Age of a GBM PatientDiagnosis Is Coincident With EnhancedGlobal ImmunosuppressionAlthough GBM is generally considered as a disease initiatedby genetic mutations, it may also arise due to immunologicaldysfunction (Shurin, 2012); although it is not clear whichevent(s) precede(s) the other in contributing to GBM cell

Frontiers in Pharmacology | www.frontiersin.org 7 February 2019 | Volume 10 | Article 200

Page 8: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 8

799

800

801

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

825

826

827

828

829

830

831

832

833

834

835

836

837

838

839

840

841

842

843

844

845

846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

888

889

890

891

892

893

894

895

896

897

898

899

900

901

902

903

904

905

906

907

908

909

910

911

912

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

initiation/outgrowth. While immunosuppression is routinelyanalyzed in GBM patients following a diagnosis (Beatty andGladney, 2015), few studies have prospectively investigatedthe correlation between progressive aging and the level ofimmunosuppression in human subjects. With the hypothesis thatthe suppression of the normal immune system contributes tothe microenvironment necessary to facilitate GBM cell initiation,the 10k Immunomes database was profiled for immune systemstatus among healthy human subjects. There were substantialchanges in the peripheral immune system associated with aging.While there were no noticeable trends among total lymphocytes(Figure 8A) and CD19+ B cell (Figure 8B) levels, analysisof T cell subpopulations revealed an interesting phenotypictrend. As demonstrated in Figure 8C, CD4+ T cell levels aremaximal in the 60–69 age range. Drilling down into the CD4+T cell subset revealed that the maximal increase was primarilyattributable to the increase of immunosuppressive regulatoryT cells (Tregs; CD3+CD4+FoxP3+) within the same 60–69age group (Figure 8D). Strikingly, the cytotoxic CD8+ T cellscoincidently decreased with progressive aging (Figure 8E). Sincea high CD8+ T/Treg ratio is associated with improved overallsurvival, (Yue et al., 2014; Shang et al., 2015) and because there isa decreased CD8+ T/Treg ratio during advanced aging of normalhuman healthy individuals that is maximal in the 60–69 agegroup, the data collectively demonstrate a trend for increasedimmunosuppression in the peripheral blood coincident with themedian age of a GBM patient diagnosis.

We previously demonstrated that immunosuppressive IDO1is significantly increased in the normal healthy brain of72–74 week old mice as compared with young 6–8 week

old counterparts; independent of tumor burden (Ladomerskyet al., 2016, 2018). To determine whether this observationis generalizable to humans, RNA-sequencing data from theGTEx database was analyzed for mRNA expression levelsof established immunoregulatory genes in normal humanbrain (Figure 9A). Similar to our analysis of increased IDO1expression levels in the brain of mice during advanced aging,IDO1 is also increased in the normal human brain amongthe 60–69 age group as compared to human subjects inthe 50–59 age group (P = 0.02; Figure 9A; SupplementaryFigure S1). Although this trend remained when comparingthe 60–69 age group to other age cohorts, it did not achievestatistical significance due to insufficient human samples forcomparison. Similar to increased IDO1 mRNA expression, geneexpression for immunosuppressive PD-L1 was also highestin the 60–69 year old age group as compared with the20–29 year old age group (P = 0.01). Unexpectedly, CD11cmRNA levels, a marker traditionally associated with immunesentinel dendritic cells, was also increased in the 60–69 agegroup as compared with human subjects in the 50–59 agegroup (P = 0.02; Figure 9A). Interestingly, samples containingthe highest IDO1 gene expression (Figure 9A; green) alsoshowed high PD-L1 and CD11c levels, possibly suggesting animmunosuppressive phenotype with dendritic cell accumulationin those samples. In contrast to the enrichment of selectimmunosuppressive factors significantly increased in the normalhuman brain, IDO1 mRNA levels do not significantly changein pancreatic, skin, or thyroid tissues across age groups(Figure 9B; Supplementary Figure S2), nor any other humantissue analyzable in the GTEx database. Collectively, these

FIGURE 8 | The peripheral blood CD8+ T/Treg ratio decreases with progressive aging in healthy human subjects. 10k immunomes CyTOF data from the peripheralblood of healthy patients analyzed for (A) total lymphocytes, (B) B cells, (C) CD4+ T cells, (D) CD8+ T cells, (E) Tregs (CD4+CD25+FoxP3+), and (F) CD8+ T/Tregratio. Data was analyzed among age groups including 20–29 (n = 110), 30–39 (n = 14), 40–49 (n = 76), 50–59 (n = 78), 60–69 (n = 72), 70–79 (n = 57), 80+(n = 127). ∗P < 0.05; ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001.

Frontiers in Pharmacology | www.frontiersin.org 8 February 2019 | Volume 10 | Article 200

Page 9: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 9

913

914

915

916

917

918

919

920

921

922

923

924

925

926

927

928

929

930

931

932

933

934

935

936

937

938

939

940

941

942

943

944

945

946

947

948

949

950

951

952

953

954

955

956

957

958

959

960

961

962

963

964

965

966

967

968

969

970

971

972

973

974

975

976

977

978

979

980

981

982

983

984

985

986

987

988

989

990

991

992

993

994

995

996

997

998

999

1000

1001

1002

1003

1004

1005

1006

1007

1008

1009

1010

1011

1012

1013

1014

1015

1016

1017

1018

1019

1020

1021

1022

1023

1024

1025

1026

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

FIGURE 9 | Gene expression for immunosuppressive and immunoregulatory factors increase in the normal human brain with aging. (A) GTEx gene expressionanalysis of IDO1, TDO2, CD3ε, PD-L1, CTLA-4, and CD11c in the brain. Data was analyzed among age groups including 20–29 (n = 38), 30–39 (n = 19), 40–49(n = 123), 50–59 (n = 386), 60–69 (n = 549), 70–79 (n = 37). Green circles across gene panels share the same sample IDs. (B) GTEx gene expression analysis ofIDO1 in the normal human pancreas (n = 167), skin (n = 812), and thyroid (n = 280). ∗P < 0.05.

data confirm that systemic and brain-specific immunoregulatoryfactors favoring immunosuppression are maximal during thetime frame associated with the median age of a GBMpatient diagnosis.

DISCUSSION

The Director of the National Cancer Institute, Dr. Normal E.Sharpless, M.D., formerly an investigator within the NationalInstitute of Aging, has highlighted the importance of therelationship between cancer and aging (Sharpless, 2018).Cancer is a disease enriched among the elderly and is oftenassociated with deficits in normal immunosurveillance (Dunnet al., 2004; Zhong et al., 2016). Aging is associated withprogressive immunological changes throughout the body,including involution of the thymus, a critical site for pre-Tcell education and development into mature naïve T cells,a shift of the circulating T cell repertoire from a naïve tomemory phenotype, an absolute decrease in the number of

naïve T and B cells, a reduction in cytokine signaling, andreduced uptake of antigens and/or microbes by dendriticcells. To further understand the immunological changes thatoccur during aging, and whether these differences providefor preventative or therapeutic applications to individualsthat will be diagnosed with malignant primary brain cancer,we comprehensively studied multiple bioinformatic databaserepositories for aging-related associations between GBMpatient incidence and mortality, changes in the peripheralimmune response of normal healthy human subjects, andgene expression throughout the normal healthy human bodyfor immunoregulatory factors. Accordingly, the scientificpremise of our investigation aimed to identify meaningfulrelationships between the ages of a GBM patient diagnosis,with differences in the immune system that are potentiallytherapeutically targetable.

During our investigation, we discovered that the United Statesrate of incidence and mortality for brain and other CNS malig-nancies has been rising over the past 40 years, and due to its

Frontiers in Pharmacology | www.frontiersin.org 9 February 2019 | Volume 10 | Article 200

Page 10: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 10

1027

1028

1029

1030

1031

1032

1033

1034

1035

1036

1037

1038

1039

1040

1041

1042

1043

1044

1045

1046

1047

1048

1049

1050

1051

1052

1053

1054

1055

1056

1057

1058

1059

1060

1061

1062

1063

1064

1065

1066

1067

1068

1069

1070

1071

1072

1073

1074

1075

1076

1077

1078

1079

1080

1081

1082

1083

1084

1085

1086

1087

1088

1089

1090

1091

1092

1093

1094

1095

1096

1097

1098

1099

1100

1101

1102

1103

1104

1105

1106

1107

1108

1109

1110

1111

1112

1113

1114

1115

1116

1117

1118

1119

1120

1121

1122

1123

1124

1125

1126

1127

1128

1129

1130

1131

1132

1133

1134

1135

1136

1137

1138

1139

1140

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

enrichment among the elderly population, may reflect theincreased number of individuals that have a higher propensityfor brain cancer diagnoses as compared with younger subjects(Figures 1, 3, 4, 5). This differs from the trend for allmalignancies, where the incidence and mortality rates declinebetween the years 1995 and 2015 (Figures 1C,D). This maybe due to an increase in the number of effective therapies fortreating other types of cancer, as well as improved preventionand detection techniques for high incidence malignanciesincluding breast cancer and melanoma. According to a 2018projection by the NCI (see text footnote 3), brain and otherCNS malignancies have the third highest mortality:incidence(MIR) ratio when comparing across major cancer subtypes,which reflects a high degree of aggressiveness. Strikingly,brain cancer and other CNS malignancies were predictedto be the most expensive cancer on a per patient basis inthe year 2018 (Figure 2). Further investigation of gliomaincidence data revealed that GBM diagnoses are highlyenriched among the elderly. When the mortality data wasaccounted for, the MIR for individuals with GBM betweenthe years 2011–2015 was 1.00 (Figure 6). Analysis of TCGAdata revealed that not only are the elderly at a higherrisk for GBM, but this cohort of individuals also havea significantly worse prognosis when diagnosed with GBM(Figure 7). Further evaluation revealed a peak GBM patientincidence/mortality rate that corresponded to the maximal levelsof immunosuppressive IDO1 and PD-L1 mRNA expressionin the CNS, as well as immunosuppressive peripheral Tregabundance (Figures 8, 9). Interestingly, the CD4+FoxP3− T cellswere less affected by aging (Figure 8). A hypothesis for theincreased immunosuppressive markers in the 60–69 age group,but not among even older age groups, is that only a subsetof individuals experience this increase. It’s also possible thatthe transient increase in local immunosuppression synergizeswith peripheral mechanisms during a timeframe of substantialhormonal imbalance (i.e., menopause). Interestingly, CD11cmRNA expression also increased in the elderly human brain,which may be associated with the accumulation of brain-resident cells (Bulloch et al., 2008; D’Agostino et al., 2012;Kaunzner et al., 2012).

Since cancer is enriched among the elderly, combinedwith the increasing size of the population with advanced age(Figures 1A,B), it was surprising to find that not all malignancieshad an associated increased mortality rate when comparingdata between 1975 and 2015. Several hypotheses may explainthe rise in mortality of primary malignant brain cancer ascompared to other malignancies including: (i) the lack of effectivetreatment options for patients diagnosed with incurable braintumors as compared to therapeutic improvements for non-CNSmalignancies; (ii) a steady enrichment in factors that promotethe development of more malignant primary brain tumors; (iii)a more aggressive natural history in established primary braintumors; (iv) an absolute increase in the number of elderlyindividuals that have a higher chance for developing primarymalignant glioma; (v) an increase over time of elderly patientsdiagnosed with malignant brain tumors due to more aggressivepatient work ups providing for a larger pool of individuals

contributing to the mortality statistics; or (vi) a combination ofall stated potential conferring factors.

The CNS is a potently immunosuppressive andimmunospecialized organ, as compared with peripheral tissues(Carson et al., 2006). Through normal human subject geneexpression analysis, we found increased immunosuppressiveIDO1 in the brain that was maximally enhanced in the60–69 year old subgroup, which is complementary to ourprevious work demonstrating increased IDO1 in the normalbrain of mice during advanced aging (Ladomersky et al., 2016,2018). Unexpectedly, IDO1 enzyme activity was not increasedin the aged brain, questioning the functionality of the increasedbrain IDO1 expression. Recent studies in our laboratory andothers have hypothesized that the immunosuppressive functionof IDO1 may be, in-part, independent of its associated enzymeactivity (Pallotta et al., 2011; Wainwright et al., 2012; Zhaiet al., 2017). Although additional studies are required to fullyunderstand the relationship between increased human brainIDO1 expression and advanced aging, a hypothesis that currentlyfits the available data is that IDO1 expression increases CNSimmunosuppression through non-enzyme activity. Whetherthe basal increase of brain IDO1 expression contributes to themicroenvironment required for GBM cell initiation is also anintriguing consideration.

Another factor for considering the effects of aging onimmunosuppression and GBM onset is the differences betweenprimary, or de novo GBM, and secondary GBM; the latter ofwhich develops from lower grade II or III glioma. PrimaryGBM represents a majority of all malignant glioma cases(>90%) (Ohgaki and Kleihues, 2007). Primary GBM is routinelyassociated with mutation of PTEN (Ohgaki et al., 2004), EGFRamplification (Ekstrand et al., 1992), and p16INK4a deletion(Biernat et al., 1997). Secondary GBM, which accounts for<10% of GBM diagnoses, tends to arise in younger individualsand is characterized by the presence of mutated isocitratedehydrogenase (mIDH) and TP53 (Kohanbash et al., 2017). Themedian age of diagnosis for patients with a secondary GBMis ∼45 years old (Ohgaki and Kleihues, 2005). The differentages at which primary and secondary GBMs arise may reflectdifferent microenvironmental CNS conditions that contribute toGBM cell initiation. Highlighting this plurality, GBM presentingwith a mIDH is often associated with immune cell exclusionand almost totally absent of tumor cell-killing cytolytic CD8+ Tcells (Kohanbash et al., 2017). Despite the minimal infiltrationof immune cells, the presence of mIDH is associated with afavorable prognosis in GBM, as compared with primary GBMthat predominantly contain wild-type IDH (Yan et al., 2009).

Our comprehensive analysis across multiple databasesprovides a unique perspective for assessing the risk of GBM inthe elderly. While the data show interesting trends, a prospectiveanalysis of a large patient cohort is warranted to validate thehypotheticals proposed by our current study. Some of theseconsiderations will be built into an upcoming prospectivephase I/II trial evaluating newly diagnosed GBM patients,before and after treatment with standard radiation, nivolumab(anti-PD-1 mAb) and BMS986205 (IDO1 enzyme inhibitor),led by our group. Further analysis of TCGA survival data

Frontiers in Pharmacology | www.frontiersin.org 10 February 2019 | Volume 10 | Article 200

Page 11: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 11

1141

1142

1143

1144

1145

1146

1147

1148

1149

1150

1151

1152

1153

1154

1155

1156

1157

1158

1159

1160

1161

1162

1163

1164

1165

1166

1167

1168

1169

1170

1171

1172

1173

1174

1175

1176

1177

1178

1179

1180

1181

1182

1183

1184

1185

1186

1187

1188

1189

1190

1191

1192

1193

1194

1195

1196

1197

1198

1199

1200

1201

1202

1203

1204

1205

1206

1207

1208

1209

1210

1211

1212

1213

1214

1215

1216

1217

1218

1219

1220

1221

1222

1223

1224

1225

1226

1227

1228

1229

1230

1231

1232

1233

1234

1235

1236

1237

1238

1239

1240

1241

1242

1243

1244

1245

1246

1247

1248

1249

1250

1251

1252

1253

1254

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

allows for a valuable comparison of prognostic criteriabetween elderly and younger individuals, but still does notaccount for major differences of tumor biology. Future studiesaimed at evaluating aging-related mechanisms and increasedimmunosuppression will allow for a validation of whether thereare therapeutically targetable changes that potentially preventbrain tumor incidence and/or enhance the effectiveness ofimmunotherapeutic treatments.

Taken together, these data suggest that immunosuppressivechanges in the brain are affected by processes mediated byaging and may contribute to the significantly increased braincancer mortality rate enriched within the elderly population.The median age of a GBM diagnosis coincides with animmunosuppressive phenotype in the peripheral blood andinside the brain parenchyma. Potentially, subsets of individualswith altered expression of immunoregulatory genes possess anenhanced risk for developing GBM. A high priority must beplaced on determining whether these gene expression changescontribute to tumor cell initiation and/or progression, as wellas the ability to detect increases in CNS immunosuppressionthrough a peripheral (i.e., outside of the CNS) biomarker. Furtherresearch in this area will not only allow for a better understandingof elderly GBM patient treatment, but also potentially contributeto the ability of identifying individuals at a higher risk fordeveloping the terminal disease.

DATA AVAILABILITY

All datasets generated for this study are included in themanuscript and/or the Supplementary Files.

AUTHOR CONTRIBUTIONS

EL performed a majority of the data mining associated withthis submitted work. Data analysis was performed by EL, withstatistical input from DS. All data were reviewed by DMS, MK,EAH, ETB, SO, LZ, KLL, JC, JAS, JDW, BZ, and RVL. EL andDAW prepared the figures and wrote the manuscript.

FUNDING

EL was supported by PHS grant number T32CA070085. DAWwas supported by PHS grant number R01NS097851 awardedby the NIH/NINDS, United States Department of Health andHuman Services. RVL and DAW are supported by PHS grantnumber P50CA221747 awarded by the NIH/NCI, United StatesDepartment of Health and Human Services; the Gail BoyterMagness Foundation; the Grace Giving Foundation. ETB wassupported by PHS grant 5R50CA221848-02 awarded by the NCI.

SUPPLEMENTARY MATERIAL

The Supplementary Material for this article can be foundonline at: https://www.frontiersin.org/articles/10.3389/fphar.2019.00200/full#supplementary-material

FIGURE S1 | GTEx Analysis. GTEx gene expression analysis of IDO1, TDO2,CD3e, and PD-L1 in the brain, pancreas, skin, and thyroid.

FIGURE S2 | GTEx Analysis. GTEx gene expression analysis of CTLA-4, CD11c,PTGS2, and SLC6A4 in the brain, pancreas, skin, and thyroid.

REFERENCESAdachi, K., and Davis, M. M. (2011). T-cell receptor ligation induces distinct

signaling pathways in naive vs. antigen-experienced T cells. Proc. Natl. Acad.Sci. U.S.A. 108, 1549–1554. doi: 10.1073/pnas.1017340108

Antonia, S. J., Lopez Martin, J. A., Bendell, J., Ott, P. A., Taylor, M., Eder, J. P.,et al. (2016). Nivolumab alone and nivolumab plus ipilimumab in recurrentsmall-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2trial. Lancet Oncol. 17, 883–895. doi: 10.1016/S1470-2045(16)30098-5

Aprahamian, T., Takemura, Y., Goukassian, D., and Walsh, K. (2008). Ageing isassociated with diminished apoptotic cell clearance in vivo. Clin. Exp. Immunol.152, 448–455. doi: 10.1111/j.1365-2249.2008.03658.x

Beatty, G. L., and Gladney, W. L. (2015). Immune escape mechanisms as a guidefor cancer immunotherapy. Clin. Cancer Res. 21, 687–692. doi: 10.1158/1078-0432.CCR-14-1860

Berger, N. A., Savvides, P., Koroukian, S. M., Kahana, E. F., Deimling, G. T., Rose,J. H., et al. (2006). Cancer in the elderly. Trans. Am. Clin. Climatol. Assoc. 117,147–55; discussion 155–156.

Biernat, W., Tohma, Y., Yonekawa, Y., Kleihues, P., and Ohgaki, H. (1997).Alterations of cell cycle regulatory genes in primary (de novo) and secondaryglioblastomas. Acta Neuropathol. 94, 303–309. doi: 10.1007/s004010050711

Bristol-Myers Squibb (2017). Bristol-Myers Squibb Announces Results fromCheckMate -143, a Phase 3 Study of Opdivo (nivolumab) in Patients withGlioblastoma Multiforme. Available at: https://news.bms.com/press-release/bmy/bristol-myers-squibb-announces-results-checkmate-143-phase-3-study-opdivo-nivoluma

Bulloch, K., Miller, M. M., Gal-Toth, J., Milner, T. A., Gottfried-Blackmore, A.,Waters, E. M., et al. (2008). CD11c/EYFP transgene illuminates a discrete

network of dendritic cells within the embryonic, neonatal, adult, and injuredmouse brain. J. Comp. Neurol. 508, 687–710. doi: 10.1002/cne.21668

Cambier, J. (2005). Immunosenescence: a problem of lymphopoiesis, homeostasis,microenvironment, and signaling. Immunol. Rev. 205, 5–6. doi: 10.1111/j.0105-2896.2005.00276.x

Carson, M. J., Doose, J. M., Melchior, B., Schmid, C. D., and Ploix, C. C. (2006).CNS immune privilege: hiding in plain sight. Immunol. Rev. 213, 48–65.doi: 10.1111/j.1600-065X.2006.00441.x

Chen, C. Z., Schaffert, S., Fragoso, R., and Loh, C. (2013). Regulation of immuneresponses and tolerance: the microRNA perspective. Immunol. Rev. 253,112–128. doi: 10.1111/imr.12060

Choi, E., Lee, S., Nhung, B. C., Suh, M., Park, B., Jun, J. K., et al. (2017). Cancermortality-to-incidence ratio as an indicator of cancer management outcomes inorganization for economic cooperation and development countries. Epidemiol.Health 39:e2017006. doi: 10.4178/epih.e2017006

Chongsathidkiet, P., Jackson, C., Koyama, S., Loebel, F., Cui, X., Farber, S. H., et al.(2018). Sequestration of T cells in bone marrow in the setting of glioblastomaand other intracranial tumors. Nat. Med. 24, 1459–1468. doi: 10.1038/s41591-018-0135-2

D’Agostino, P. M., Gottfried-Blackmore, A., Anandasabapathy, N., and Bulloch, K.(2012). Brain dendritic cells: biology and pathology. Acta Neuropathol. 124,599–614. doi: 10.1007/s00401-012-1018-0

Davila, D. R., Edwards, C. K. III, Arkins, S., Simon, J., and Kelley, K. W. (1990).Interferon-gamma-induced priming for secretion of superoxide anion andtumor necrosis factor-alpha declines in macrophages from aged rats. FASEB J.4, 2906–2911. doi: 10.1096/fasebj.4.11.2165948

Dunn, G. P., Old, L. J., and Schreiber, R. D. (2004). The immunobiology of cancerimmunosurveillance and immunoediting. Immunity 21, 137–148. doi: 10.1016/j.immuni.2004.07.017

Frontiers in Pharmacology | www.frontiersin.org 11 February 2019 | Volume 10 | Article 200

Page 12: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 12

1255

1256

1257

1258

1259

1260

1261

1262

1263

1264

1265

1266

1267

1268

1269

1270

1271

1272

1273

1274

1275

1276

1277

1278

1279

1280

1281

1282

1283

1284

1285

1286

1287

1288

1289

1290

1291

1292

1293

1294

1295

1296

1297

1298

1299

1300

1301

1302

1303

1304

1305

1306

1307

1308

1309

1310

1311

1312

1313

1314

1315

1316

1317

1318

1319

1320

1321

1322

1323

1324

1325

1326

1327

1328

1329

1330

1331

1332

1333

1334

1335

1336

1337

1338

1339

1340

1341

1342

1343

1344

1345

1346

1347

1348

1349

1350

1351

1352

1353

1354

1355

1356

1357

1358

1359

1360

1361

1362

1363

1364

1365

1366

1367

1368

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

Dutta, S., and Sengupta, P. (2016). Men and mice: relating their ages. Life Sci. 152,244–248. doi: 10.1016/j.lfs.2015.10.025

Ekstrand, A. J., Sugawa, N., James, C. D., and Collins, V. P. (1992). Amplified andrearranged epidermal growth factor receptor genes in human glioblastomasreveal deletions of sequences encoding portions of the N- and/or C-terminaltails. Proc. Natl. Acad. Sci. U.S.A. 89, 4309–4313. doi: 10.1073/pnas.89.10.4309

Filley, A. C., Henriquez, M., and Dey, M. (2017). Recurrent glioma clinicaltrial, CheckMate-143: the game is not over yet. Oncotarget 8, 91779–91794.doi: 10.18632/oncotarget.21586

Fox, J. G., Davisson, M. T., and Smith, A. L. (2007). The Mouse in BiomedicalResearch. New York, NY: Academic Press.

Gon, Y., Hashimoto, S., Hayashi, S., Koura, T., Matsumoto, K., and Horie, T.(1996). Lower serum concentrations of cytokines in elderly patients withpneumonia and the impaired production of cytokines by peripheral bloodmonocytes in the elderly. Clin. Exp. Immunol. 106, 120–126.

Johnson, D. R., and O’Neill, B. P. (2012). Glioblastoma survival in the United Statesbefore and during the temozolomide era. J. Neurooncol. 107, 359–364.doi: 10.1007/s11060-011-0749-4

Kaunzner, U. W., Miller, M. M., Gottfried-Blackmore, A., Gal-Toth, J., Felger,J. C., McEwen, B. S., et al. (2012). Accumulation of resident and peripheraldendritic cells in the aging CNS. Neurobiol. Aging 33, 681.e1–693.e1. doi: 10.1016/j.neurobiolaging.2010.06.007

Kohanbash, G., Carrera, D. A., Shrivastav, S., Ahn, B. J., Jahan, N., and Mazor, T.(2017). Isocitrate dehydrogenase mutations suppress STAT1 and CD8+ T cellaccumulation in gliomas. J. Clin. Invest. 127, 1425–1437. doi: 10.1172/JCI90644

Kugel, C. H. III, Douglass, S. M., Webster, M. R., Kaur, A., Liu, Q., Yin, X.,et al. (2018). Age correlates with response to anti-PD1, reflecting age-relateddifferences in intratumoral effector and regulatory T-cell populations. Clin.Cancer Res. 24, 5347–5356. doi: 10.1158/1078-0432.CCR-18-1116

Ladomersky, E., Zhai, L., Gritsina, G., Genet, M., Lauing, K. L., Wu, M.,et al. (2016). Wainwright, advanced age negatively impacts survival in anexperimental brain tumor model. Neurosci. Lett. 630, 203–208. doi: 10.1016/j.neulet.2016.08.002

Ladomersky, E., Zhai, L., Lenzen, A., Lauing, K. L., Qian, J., Scholtens, D. M.,et al. (2018). IDO1 inhibition synergizes with radiation and PD-1 blockade todurably increase survival against advanced Glioblastoma. Clin. Cancer Res. 24,2559–2573. doi: 10.1158/1078-0432.CCR-17-3573

Larkin, J., Chiarion-Sileni, V., Gonzalez, R., Grob, J. J., Cowey, C. L., Lao,C. D., et al. (2015). Combined nivolumab and ipilimumab or monotherapy inuntreated melanoma.N. Engl. J. Med. 373, 23–34. doi: 10.1056/NEJMoa1504030

Li, G., Yu, M., Lee, W. W., Tsang, M., Krishnan, E., Weyand, C. M., et al. (2012).Decline in miR-181a expression with age impairs T cell receptor sensitivity byincreasing DUSP6 activity. Nat. Med. 18, 1518–1524. doi: 10.1038/nm.2963

Liu, Y., Sanoff, H. K., Cho, H., Burd, C. E., Torrice, C., Ibrahim, J. G., et al. (2009).Expression of p16(INK4a) in peripheral blood T-cells is a biomarker of humanaging. Aging Cell 8, 439–448. doi: 10.1111/j.1474-9726.2009.00489.x

McElhaney, J. E., and Dutz, J. P. (2008). Better influenza vaccines for older people:what will it take? J. Infect. Dis. 198, 632–634. doi: 10.1086/590435

Motzer, R. J., Escudier, B., McDermott, D. F., George, S., Hammers, H. J.,Srinivas, S., et al. (2015). Nivolumab versus everolimus in advanced renal-cellcarcinoma. N. Engl. J. Med. 373, 1803–1813. doi: 10.1056/NEJMoa1510665

Nduom, E. K., Wei, J., Yaghi, N. K., Huang, N., Kong, L. Y., Gabrusiewicz, K., et al.(2016). PD-L1 expression and prognostic impact in glioblastoma. Neuro Oncol.18, 195–205. doi: 10.1093/neuonc/nov172

Nikolich-Zugich, J. (2018). The twilight of immunity: emerging concepts in agingof the immune system. Nat. Immunol. 19, 10–19. doi: 10.1038/s41590-017-0006-x

Ohgaki, H., Dessen, P., Jourde, B., Horstmann, S., Nishikawa, T., Di Patre, L. P.,et al. (2004). Genetic pathways to glioblastoma: a population-based study.Cancer Res. 64, 6892–6899. doi: 10.1158/0008-5472.CAN-04-1337

Ohgaki, H., and Kleihues, P. (2005). Population-based studies on incidence,survival rates, and genetic alterations in astrocytic and oligodendroglialgliomas. J. Neuropathol. Exp. Neurol. 64, 479–489. doi: 10.1093/jnen/64.6.479

Ohgaki, H., and Kleihues, P. (2007). Genetic pathways to primary and secondaryglioblastoma. Am. J. Pathol. 170, 1445–1453. doi: 10.2353/ajpath.2007.070011

Pallotta, M. T., Orabona, C., Volpi, C., Vacca, C., Belladonna, M. L., Bianchi, R.,et al. (2011). Indoleamine 2,3-dioxygenase is a signaling protein in long-termtolerance by dendritic cells. Nat. Immunol. 12, 870–878. doi: 10.1038/ni.2077

Pang, W. W., Price, E. A., Sahoo, D., Beerman, I., Maloney, W. J., Rossi, D. J.,et al. (2011). Human bone marrow hematopoietic stem cells are increasedin frequency and myeloid-biased with age. Proc. Natl. Acad. Sci. U.S.A. 108,20012–20017. doi: 10.1073/pnas.1116110108

Perry, V. H., Matyszak, M. K., and Fearn, S. (1993). Altered antigen expression ofmicroglia in the aged rodent CNS. Glia 7, 60–67. doi: 10.1002/glia.440070111

Ries, L. A. G., Harkins, D., Krapcho, M., Mariotto, A., Miller, B. A., Feuer, E. J.,et al. (2006). SEER Cancer Statistics Review, 1975-2003. Behtesda, MD: NationalCancer Institute.

Rogers, J., Luber-Narod, J., Styren, S. D., and Civin, W. H. (1988). Expressionof immune system-associated antigens by cells of the human central nervoussystem: relationship to the pathology of Alzheimer’s disease. Neurobiol. Aging9, 339–349. doi: 10.1016/S0197-4580(88)80079-4

Rossi, D. J., Bryder, D., Zahn, J. M., Ahlenius, H., Sonu, R., Wagers, A. J., et al.(2005). Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc.Natl. Acad. Sci. U.S.A. 102, 9194–9199. doi: 10.1073/pnas.0503280102

Schreiber, K., Arina, A., Engels, B., Spiotto, M. T., Sidney, J., Sette, A., et al. (2012).Spleen cells from young but not old immunized mice eradicate large establishedcancers.Clin. Cancer Res. 18, 2526–2533. doi: 10.1158/1078-0432.CCR-12-0127

Shang, B., Liu, Y., Jiang, S. J., and Liu, Y. (2015). Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review andmeta-analysis. Sci. Rep. 5:15179. doi: 10.1038/srep15179

Sharpless, N. (2018). The Challenging Landscape of Cancer and Aging: Chartinga Way Forward. Available at: https://www.cancer.gov/news-events/cancer-currents-blog/2018/sharpless-aging-cancer-research

Shurin, M. R. (2012). Cancer as an immune-mediated disease. Immunotargets Ther.1, 1–6. doi: 10.2147/ITT.S29834

Stupp, R., Hegi, M. E., Mason, W. P., van den Bent, M. J., Taphoorn, M. J., Janzer,R. C., et al. (2009). Effects of radiotherapy with concomitant and adjuvanttemozolomide versus radiotherapy alone on survival in glioblastoma in arandomised phase III study: 5-year analysis of the EORTC-NCIC trial. LancetOncol. 10, 459–466. doi: 10.1016/S1470-2045(09)70025-7

Stupp, R., Mason, W. P., van den Bent, M. J., Weller, M., Fisher, B., Taphoorn,M. J., et al. (2005). Radiotherapy plus concomitant and adjuvant temozolomidefor glioblastoma. N. Engl. J. Med. 352, 987–996. doi: 10.1056/NEJMoa043330

Stupp, R., Taillibert, S., Kanner, A., Read, W., Steinberg, D., Lhermitte, B., et al.(2017). Effect of tumor-treating fields plus maintenance temozolomide vsmaintenance temozolomide alone on survival in patients with glioblastoma:a randomized clinical trial. JAMA 318, 2306–2316. doi: 10.1001/jama.2017.18718

Wainwright, D. A., Balyasnikova, I. V., Chang, A. L., Ahmed, A. U., Moon,K. S., Auffinger, B., et al. (2012). IDO expression in brain tumors increases therecruitment of regulatory T cells and negatively impacts survival. Clin. CancerRes. 18, 6110–6121. doi: 10.1158/1078-0432.CCR-12-2130

Wen, P. Y., and Kesari, S. (2008). Malignant gliomas in adults. N. Engl. J. Med. 359,492–507. doi: 10.1056/NEJMra0708126

Wick, G., Backovic, A., Rabensteiner, E., Plank, N., Schwentner, C., and Sgonc, R.(2010). The immunology of fibrosis: innate and adaptive responses. TrendsImmunol. 31, 110–119. doi: 10.1016/j.it.2009.12.001

Yan, H., Parsons, D. W., Jin, G., McLendon, R., Rasheed, B. A., Yuan, W., et al.(2009). IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765–773.doi: 10.1056/NEJMoa0808710

Yancik, R., Wesley, M. N., Ries, L. A., Havlik, R. J., Edwards, B. K., and Yates, J. W.(2001). Effect of age and comorbidity in postmenopausal breast cancer patientsaged 55 years and older. JAMA 285, 885–892. doi: 10.1001/jama.285.7.885

Yancik, R., Wesley, M. N., Ries, L. A., Havlik, R. J., Long, S., Edwards, B. K., et al.(1998). Comorbidity and age as predictors of risk for early mortality of maleand female colon carcinoma patients: a population-based study. Cancer 82,2123–2134. doi: 10.1002/(SICI)1097-0142(19980601)82:11<2123::AID-CNCR6>3.0.CO;2-W

Young, J. S., Chmura, S. J., Wainwright, D. A., Yamini, B., Peters, K. B., and Lukas,R. V. (2017). Management of glioblastoma in elderly patients. J. Neurol. Sci. 380,250–255. doi: 10.1016/j.jns.2017.07.048

Yue, Q., Zhang, X., Ye, H. X., Wang, Y., Du, Z. G., Yao, Y., et al. (2014). Theprognostic value of Foxp3+ tumor-infiltrating lymphocytes in patients withglioblastoma. J. Neurooncol. 116, 251–259. doi: 10.1007/s11060-013-1314-0

Zeng, D. Q., Yu, Y. F., Ou, Q. Y., Li, X. Y., Zhong, R. Z., Xie, C. M., et al. (2016).Prognostic and predictive value of tumor-infiltrating lymphocytes for clinical

Frontiers in Pharmacology | www.frontiersin.org 12 February 2019 | Volume 10 | Article 200

Page 13: The Coincidence Between Increasing Age, Immunosuppression, … · 2020. 10. 1. · (Wick et al., 2010). T cell senescence increases with progressive aging through the induction of

fphar-10-00200 March 19, 2019 Time: 19:10 # 13

1369

1370

1371

1372

1373

1374

1375

1376

1377

1378

1379

1380

1381

1382

1383

1384

1385

1386

1387

1388

1389

1390

1391

1392

1393

1394

1395

1396

1397

1398

1399

1400

1401

1402

1403

1404

1405

1406

1407

1408

1409

1410

1411

1412

1413

1414

1415

1416

1417

1418

1419

1420

1421

1422

1423

1424

1425

1426

1427

1428

1429

1430

1431

1432

1433

1434

1435

1436

1437

1438

1439

1440

1441

1442

1443

1444

1445

1446

1447

1448

1449

1450

1451

1452

1453

1454

1455

1456

1457

1458

1459

1460

1461

1462

1463

1464

1465

1466

1467

1468

1469

1470

1471

1472

1473

1474

1475

1476

1477

1478

1479

1480

1481

1482

Ladomersky et al. Aging, Immunosuppression, and Malignant Glioma

therapeutic research in patients with non-small cell lung cancer. Oncotarget 7,13765–13781. doi: 10.18632/oncotarget.7282

Zhai, L., Ladomersky, E., Lauing, K. L., Wu, M., Genet, M., and Gritsina, G. (2017).Infiltrating T cells increase IDO1 expression in glioblastoma and contribute todecreased patient survival. Clin. Cancer Res. 23, 6650–6660. doi: 10.1158/1078-0432.CCR-17-0120

Zhai, L., Ladomersky, E., Lenzen, A., Nguyen, B., Patel, R., Lauing, K. L., et al.(2018). Wainwright, IDO1 in cancer: a Gemini of immune checkpoints. Cell.Mol. Immunol. 15, 447–457. doi: 10.1038/cmi.2017.143

Zhong, Z., Sanchez-Lopez, E., and Karin, M. (2016). Autophagy, inflammation,and immunity: a troika governing cancer and its treatment. Cell 166, 288–298.doi: 10.1016/j.cell.2016.05.051

Conflict of Interest Statement: The authors declare that the research wasconducted in the absence of any commercial or financial relationships that couldbe construed as a potential conflict of interest.

Copyright © 2019 Ladomersky, Scholtens, Kocherginsky, Hibler, Bartom, Otto-Meyer, Zhai, Lauing, Choi, Sosman, Wu, Zhang, Lukas and Wainwright. This is anopen-access article distributed under the terms of the Creative Commons AttributionLicense (CC BY). The use, distribution or reproduction in other forums is permitted,provided the original author(s) and the copyright owner(s) are credited and that theoriginal publication in this journal is cited, in accordance with accepted academicpractice. No use, distribution or reproduction is permitted which does not complywith these terms.

Frontiers in Pharmacology | www.frontiersin.org 13 February 2019 | Volume 10 | Article 200